C4BPB Human

Complement Component 4 Binding Protein, Beta Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to C4BPB Human

C4BPB (Complement Component 4 Binding Protein Beta) is a critical regulatory protein in the human immune system, primarily functioning as the beta-chain component of the C4b-binding protein (C4BP). It plays a central role in modulating complement activation, coagulation, and inflammation. Structurally, C4BPB assembles with seven identical alpha-chains to form the major isoform C4BP(β+), which binds anticoagulant protein S and regulates immune responses .

Functional Roles

C4BPB’s beta-chain is essential for interactions with protein S, a vitamin K-dependent anticoagulant, and contributes to C4BP’s dual roles in complement regulation and coagulation .

Canonical Functions

  • Complement Inhibition:

    • Accelerates decay of C3 convertase (C4bC2a) by dissociating C2a .

    • Acts as a cofactor for factor I-mediated cleavage of C4b and C3b .

  • Coagulation Regulation:

    • Binds protein S, enhancing activated protein C’s anticoagulant activity .

Non-Canonical Functions

  • Inflammation Modulation:

    • C4BP(β-) (lacking the beta-chain) exhibits anti-inflammatory effects by downregulating pro-inflammatory cytokines (e.g., TNF-α, IL-12) and upregulating IL-10 .

  • Pathogen Resistance:

    • Binds viral spike proteins (e.g., SARS-CoV-2), inhibiting viral transduction .

    • Acts as an entry inhibitor for H1N1 influenza but promotes H3N2 entry, depending on viral subtype .

Isoforms and Physiological Relevance

C4BP exists as two major isoforms:

IsoformCompositionFunction
C4BP(β+)7 α-chains + 1 β-chainPrimary complement inhibitor; binds protein S for coagulation regulation .
C4BP(β-)7 α-chainsAnti-inflammatory; upregulated during acute inflammation (e.g., SLE flares) .

Under inflammatory conditions, the ratio of C4BP(β+):C4BP(β-) shifts from 80:20 to 50:50, enhancing immunomodulatory activity .

Disease Associations

DiseaseMechanism
Systemic Lupus Erythematosus (SLE)C4BP(β-) reduces pro-inflammatory cytokine secretion and kidney inflammation .
InfectionsPathogens (e.g., Streptococcus pyogenes) recruit C4BP to evade complement .
CancerPromotes tumor survival by limiting complement-mediated cytotoxicity .

Therapeutic Potential

  • Recombinant C4BP Variants:

    • PRP6-HO7: A minimal recombinant variant (CCP6 + oligomerization domain) mimics C4BP(β-)'s anti-inflammatory effects without pathogen-binding risks .

    • C4BP-IgM Fusion: Neutralizes pathogens like Neisseria gonorrhoeae by blocking C4BP recruitment .

Viral Entry Modulation

C4BP’s α-chain binds influenza hemagglutinin (HA), neuraminidase (NA), and matrix protein 1 (MP1) :

Viral SubtypeC4BP EffectMechanism
H1N1Inhibits entry; suppresses replicationBinds HA/NA via CCP domains 1-2, 4-5, 7-8 .
H3N2Promotes entry; enhances replicationWeak binding to HA/NA; pro-inflammatory response .

Autoimmune Disease Interventions

In SLE, C4BP(β-) reduces dendritic cell maturation (CD83, CD86) and cytokine secretion (IL-12, TNF-α), while C4BP(β+)-PS lacks this activity .

Future Directions

  1. Therapeutic Engineering:

    • Designing CCP6-based multimers for SLE treatment .

    • Targeting C4BP-pathogen interactions to prevent immune evasion .

  2. Complement-Independent Roles:

    • Exploring C4BP’s interaction with extracellular DNA, CD40, and heparin in autoimmune and clotting disorders .

Product Specs

Introduction
C4BPB (Complement Component 4 Binding Protein, Beta) is a protein that plays a crucial role in regulating the complement system, a part of the immune system. It belongs to a family of proteins characterized by repeating structural units. C4BPB primarily functions by inhibiting the complement cascade, preventing excessive activation that can damage the body's own tissues. It achieves this by acting as a cofactor for C3bINA, an enzyme that inactivates complement components C3b and C4b. Moreover, C4BPB facilitates the breakdown of the C4bC2a complex, further controlling complement activation. Additionally, C4BPB interacts with Protein S, a protein involved in blood clotting regulation, and serum amyloid P component, highlighting its diverse roles in immune and coagulation systems.
Description
This product consists of a recombinant human C4BPB protein produced in E. coli bacteria. It is a single, non-glycosylated polypeptide chain, meaning it lacks attached sugar molecules. The protein contains 260 amino acids, with a molecular weight of 29kDa. For purification and detection purposes, a 25 amino acid His-tag is attached to the N-terminus of the protein. The purification process utilizes proprietary chromatographic techniques to ensure high purity.
Physical Appearance
Clear, colorless solution that has been sterilized by filtration.
Formulation
The C4BPB protein is provided in a solution with a concentration of 1mg/ml. The solution contains 20mM Tris-HCl buffer at pH 8.0, 10% glycerol, and 0.15M NaCl. These components help maintain the protein's stability and activity.
Stability
For short-term storage (up to 4 weeks), the product can be stored at 4°C. For longer-term storage, it is recommended to store the product frozen at -20°C. To further enhance stability during long-term storage, consider adding a carrier protein such as HSA or BSA at a concentration of 0.1%. It's important to avoid repeated freezing and thawing of the product to prevent protein degradation.
Purity
The purity of the C4BPB protein is greater than 90%, as determined by SDS-PAGE analysis, a widely used technique for assessing protein purity.
Synonyms
C4b-binding protein beta chain, C4BPB, C4BP.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSHMSDAEH CPELPPVDNS IFVAKEVEGQ ILGTYVCIKG YHLVGKKTLF CNASKEWDNT TTECRLGHCP DPVLVNGEFS SSGPVNVSDK ITFMCNDHYI LKGSNRSQCL EDHTWAPPFP ICKSRDCDPP GNPVHGYFEG NNFTLGSTIS YYCEDRYYLV GVQEQQCVDG EWSSALPVCK LIQEAPKPEC EKALLAFQES KNLCEAMENF MQQLKESGMT MEELKYSLEL KKAELKAKLL.

Q&A

What is the biological function of C4BPB in the coagulation cascade?

C4BPB (C4b-binding protein Beta chain) plays a crucial role in the coagulation/fibrinolysis cascade primarily through its binding with protein S (PS), a key anticoagulant protein. The β-chain of C4BP is specific for binding protein S, which influences the regulation of thrombin generation .

These complex interactions explain why, despite protein S's key role in regulating thrombin generation, its involvement in venous thrombosis (VT) susceptibility remains incompletely understood. While protein S deficiency is considered a risk factor for VT, total and free protein S levels show limited correlation with thrombotic events .

What are the different isoforms of C4BP and how does the β chain contribute to their structure?

C4BP exists in three primary isoforms in plasma:

  • α7β1 (main isoform)

  • α7β0 (approximately 17% of circulating C4BP)

  • α6β1

These isoforms differ based on the number of identical α-chains (6 or 7) and the presence or absence of a single β-chain . While the α-chains can bind various ligands, only the β-chain specifically binds protein S . Consequently, only isoforms containing the β-chain (α7β1 and α6β1) can form complexes with protein S.

Plasma levels of all three isoforms have high heritability estimates (approximately 40% each) , but our understanding of their genetic determinants remained limited until recent genome-wide association studies focused on the C4BPA and C4BPB genes in chromosome 1q32 .

What methodologies are used to measure C4BP isoforms in clinical research?

Measurement of C4BP isoforms typically employs mouse anti-human antibodies specific to the α and β chains. Researchers obtain plasma levels of C4BPα and C4BPβ using these antibodies . The percentage of C4BP β-chain lacking isoform (%α7β0) is calculated from the ratio (C4BPα − C4BPβ)/C4BPα, which effectively measures the molar excess of C4BPα-chain .

In clinical research settings, these measurements are often complemented by enzyme-linked immunosorbent assays (ELISAs) to measure free protein S (fPS) and total protein S levels, providing a more comprehensive picture of C4BP's potential influence on coagulation .

The following table shows representative measurements of C4BP isoforms in relation to different genotypes:

Gene/SNP GenotypeC4BPαC4BPβ%α7β0fPSTotal PS
C4BPA rs11120218
GG (n=235)315.1252.918.597.5101.4
GA (n=112)341.9255.624.294.7101.4
AA (n=5)383.8244.735.393.097.4
R², %3.810.0110.620.410.00
P-value5.64×10⁻⁴.89791.55×10⁻⁹.1999.5523

This methodological approach allows researchers to quantify the distribution of different C4BP isoforms and assess their relationship to genetic variants and disease risk .

How do genetic variations affect C4BPB/C4BPA expression at the mRNA and protein levels?

Genetic variations in the C4BPB/C4BPA locus affect both mRNA expression and plasma protein levels in complex ways:

At the mRNA level, the Gutenberg Heart Study examined C4BPA and C4BPB expression in monocytes from 1,490 participants . Four SNPs showed strong associations (P < 10⁻¹¹) with C4BPA expression, while showing minimal association with C4BPB expression . The SNP most strongly associated with C4BPA monocyte expression was rs11120211 (R² = 10.7%, P = 6.7 × 10⁻³⁶), with the rs11120211-A allele linked to higher expression levels in an additive model .

At the protein level, the GAIT study found that specific SNPs significantly affected plasma concentrations of C4BP isoforms:

  • The rs3813948 SNP in C4BPB explained approximately 11% of the variability in %α7β0 levels (P = 4.37×10⁻¹⁰)

  • The rs11120218 SNP in C4BPA explained approximately 3.8% of C4BPα variability (P = 5.64×10⁻⁴) and 10.6% of %α7β0 variability (P = 1.55×10⁻⁹)

Interestingly, these genetic variations did not significantly affect free or total protein S levels, suggesting that alterations in C4BP isoform composition operate through mechanisms independent of protein S binding capacity .

What evidence links C4BPB/C4BPA to venous thrombosis risk?

Compelling evidence establishes C4BPB/C4BPA as a susceptibility locus for venous thrombosis (VT) through a protein S-independent mechanism:

In a genome-wide association study (GAIT) followed by validation in two independent case-control studies (MARTHA and FARIVE), researchers found that genetic variants associated with increased α7β0 plasma levels and increased C4BPA expression were also associated with increased VT risk (odds ratio [OR] = 1.24 [1.03-1.53]) . These studies collectively included 1,706 VT cases and 1,379 controls, providing robust statistical support for the association .

Critically, the SNPs associated with VT risk showed no significant association with free protein S or total protein S levels . This finding represents a paradigm shift, as it suggests C4BPB/C4BPA influences thrombosis risk through mechanisms independent of its canonical role in protein S binding.

The working hypothesis emerging from these studies is that genetic variants affect the relative abundance of different C4BP isoforms, particularly increasing the proportion of the β-chain lacking isoform (α7β0), which may have downstream effects on coagulation through novel pathways yet to be fully characterized .

How is C4BPB implicated in neurological disorders and immune system regulation?

Recent proteomic profiling studies have revealed potential roles for C4BPB in neurological disorders through immunoregulatory mechanisms:

In a study of temporal lobe epilepsy (TLE), researchers identified C4BPB among proteins involved in multiple biological processes relevant to neurological function:

  • Immune response regulation:

    • C4BPB was identified among 14 proteins involved in "Regulation of the immune system process"

    • It was also one of 7 proteins involved in "Activation of immune response"

  • Connection to neuroinflammation:
    For both MRI-positive and MRI-negative TLE groups, overrepresented proteins (including C4BPB) were involved in immune response regulation, which is characteristic of neuroinflammatory processes . Neuroinflammation is considered one of the leading mechanisms of epileptogenesis .

  • Blood-brain barrier implications:
    The study noted that prolonged seizures associate with increased blood-brain barrier permeability , potentially explaining how plasma proteins like C4BPB might interact with central nervous system tissues in neurological disorders.

The study also found that blood immunological inflammatory substrates play roles in various neurological disorders, with evidence for the induction of plasma inflammatory markers . This was observed in both animal models of temporal lobe epilepsy and human studies .

These findings suggest C4BPB may have previously unrecognized functions in neurological disorders through inflammatory and immune-related mechanisms, extending its biological significance beyond the coagulation system.

What methodological approaches are optimal for investigating C4BPB's protein-protein interactions beyond protein S?

While C4BPB is primarily known for its protein S binding role, investigating its broader interactome requires specialized methodological approaches:

  • Affinity-based proteomics: Using purified C4BPB as bait in pull-down assays followed by mass spectrometry can identify previously unknown binding partners . This approach could reveal interactions that explain C4BPB's protein S-independent effects on thrombosis risk and potential neurological functions.

  • Structural biology techniques: X-ray crystallography or cryo-electron microscopy of C4BPB complexes could elucidate binding interfaces and interaction mechanisms. The search results indicate complex binding dynamics between C4BP and its partners that would benefit from structural characterization .

  • Functional validation assays: After identifying potential interactions, researchers should employ cell-based assays to validate these interactions under physiological conditions. For example, investigating how C4BPB affects neuroinflammatory processes in neuronal cell models could clarify its role in epileptogenesis .

  • Isoform-specific analysis: Given that C4BP exists in multiple isoforms, researchers should develop methods to study protein-protein interactions in an isoform-specific manner . This is particularly important since genetic variants affect the relative proportions of these isoforms.

  • Systems biology integration: Combining interaction data with gene expression profiles across tissues can help contextualize C4BPB's role in different physiological systems, particularly important given its potential involvement in both coagulation and neurological disorders .

These approaches would help elucidate the functional significance of C4BPB beyond its established role in protein S binding, potentially explaining its association with thrombosis risk and neurological disorders.

How can researchers differentiate between direct and indirect effects of C4BPB genetic variants on disease risk?

Differentiating between direct and indirect effects of C4BPB genetic variants on disease risk requires sophisticated study designs:

  • Mendelian randomization studies: Using C4BPB genetic variants as instrumental variables can help establish causal relationships between C4BP isoforms and disease outcomes. This approach could clarify whether the association with venous thrombosis is mediated directly by C4BPB or through other pathways .

  • Mediation analysis: Statistical approaches to quantify how much of the genetic effect on disease risk is mediated through specific biomarkers (e.g., C4BP isoform levels) versus other pathways. The search results indicate that C4BPB/C4BPA variants affect thrombosis risk independently of protein S levels, suggesting additional mediating factors exist .

  • Conditional knockouts and tissue-specific models: Animal models with conditional or tissue-specific C4BPB knockout could help isolate direct effects of C4BPB deficiency in specific tissues relevant to thrombosis or neurological disorders .

  • Multi-omics integration: Combining genomics, transcriptomics, and proteomics data can help trace the causal chain from genetic variants to disease. For example, identifying how SNPs in the C4BPB/C4BPA locus affect the transcriptome and proteome in relevant tissues could reveal the mechanistic pathways linking these variants to disease risk .

  • Functional genomics: CRISPR-based approaches to introduce specific C4BPB/C4BPA variants into cellular models could help establish direct causality between genetic variants and cellular phenotypes relevant to disease mechanisms .

These methodological approaches would help researchers move beyond association studies to establish mechanistic links between C4BPB genetic variants and disease risk, potentially identifying new therapeutic targets.

How does C4BPB contribute to neuroinflammatory processes in epilepsy and other neurological disorders?

Emerging evidence suggests C4BPB may play significant roles in neuroinflammatory processes relevant to epilepsy:

Proteomic profiling of blood plasma from temporal lobe epilepsy (TLE) patients revealed differential expression of multiple proteins involved in immune response pathways . Gene ontology analysis identified C4BPB among proteins participating in key immunoregulatory functions:

Number of Involved ProteinsHigh-Level Gene Ontology CategoryProteins Including C4BPB
14Regulation of the immune system processCPB2, HPX, C9, APOA1, CLU, C4BPB, SERPING1, PRDX2, FGG, A2M, CPN2, SELL, APOD, ORM1
7Activation of immune responseCPB2, C9, CLU, C4BPB, SERPING1, A2M, CPN2

These functional classifications highlight C4BPB's involvement in immune processes that may contribute to neuroinflammation . The study noted that neuroinflammation is one of the leading mechanisms underlying epileptogenesis, and the protein expression profiles differed between MRI-positive and MRI-negative patient groups .

The connection between peripheral immune factors and central nervous system disorders appears increasingly relevant, as studies have demonstrated that activation of the peripheral immune system can have epileptogenic effects . The search results also noted that prolonged seizures associate with increased blood-brain barrier permeability, potentially allowing plasma proteins like C4BPB to interact with brain tissue .

These findings suggest that C4BPB may contribute to neurological disorders through immune-mediated mechanisms beyond its established role in the coagulation system, opening new avenues for investigating its potential as a biomarker or therapeutic target in epilepsy.

What recent technological advances have improved our ability to study C4BPB's role in complex disorders?

Recent technological advances have significantly enhanced researchers' ability to investigate C4BPB's multifaceted roles:

  • Advanced proteomics approaches: Quantitative proteomic analysis offers unbiased screening of protein changes across conditions. The search results describe plasma proteomic studies providing information on potential biomarkers for neurological disorders . These technologies can now detect subtle alterations in C4BPB isoform composition that may be relevant to disease mechanisms.

  • Genome-wide association studies with increased resolution: The ability to analyze hundreds of thousands of SNPs across the genome has enabled researchers to identify specific C4BPB/C4BPA variants associated with protein levels and disease risk . The GAIT study analyzed 283,437 SNPs to identify genetic determinants of C4BP isoform levels .

  • Integrative multi-omics platforms: Technologies that integrate genomic, transcriptomic, and proteomic data allow researchers to trace the effects of genetic variants through biological systems. The search results demonstrate how researchers combined SNP genotyping with expression analysis and protein quantification to understand C4BPB's role in disease .

  • Sophisticated statistical methods for genetic analysis: Advanced statistical approaches like measured genotype association analysis assuming additive allele effects have improved detection of genetic associations. The SOLAR software used in the GAIT study represents this methodological advancement .

  • Improved blood-brain barrier models: Better in vitro models of the blood-brain barrier help researchers study how plasma proteins like C4BPB may interact with the central nervous system during neurological disorders .

These technological advances have transformed our ability to study complex proteins like C4BPB, revealing its potential involvement in multiple biological processes beyond its canonical role in coagulation, including immune regulation and neurological disorders.

Product Science Overview

Structure and Function

C4BPB is a component of the C4b-binding protein (C4BP), which is a large glycoprotein composed of seven identical alpha-chains and one unique beta-chain . The beta-chain of C4BP binds to protein S, a vitamin K-dependent protein that serves as a cofactor for activated protein C, which is involved in the regulation of blood coagulation . This interaction highlights the dual role of C4BP in both the complement system and the coagulation system.

The primary function of C4BP is to regulate the classical pathway of complement activation. It acts as a cofactor for the enzyme C3b/C4b inactivator (C3bINA), which hydrolyzes the complement fragment C4b . Additionally, C4BP accelerates the degradation of the C4bC2a complex (C3 convertase) by dissociating the complement fragment C2a . This regulatory role is crucial in preventing the over-activation of the complement system, which can lead to tissue damage and inflammation.

Clinical Significance

Mutations or dysregulation of the C4BPB gene can be associated with various diseases. For instance, Hemolytic Uremic Syndrome (HUS) and Atypical Hemolytic-Uremic Syndrome (aHUS) are linked to abnormalities in the complement system, including C4BPB . These conditions are characterized by the destruction of red blood cells, kidney failure, and low platelet count.

Moreover, C4BPB has been studied for its potential role as a biomarker. For example, the alpha-chain of C4BP (C4BPα) has been suggested as a biomarker to predict clopidogrel resistance in patients with coronary artery disease . Elevated levels of C4BPα in plasma may indicate a higher sensitivity to clopidogrel, a medication used to prevent blood clots .

Recombinant C4BPB

Recombinant human C4BPB is produced using recombinant DNA technology, which involves inserting the gene encoding C4BPB into a suitable expression system, such as Escherichia coli (E. coli). The recombinant protein is then purified using conventional chromatography techniques . This recombinant form is used in various research applications to study the function and regulation of the complement system and its role in disease.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.